We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

An updated review of teriflunomide's use in multiple sclerosis

    Aaron E Miller

    *Author for correspondence: Tel.: +1 212 241 6854;

    E-mail Address: aaron.miller@mssm.edu

    Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA

    Published Online:https://doi.org/10.2217/nmt-2021-0014

    Abstract

    Teriflunomide, a once daily, oral disease-modifying therapy, has demonstrated consistent efficacy, safety and tolerability in patients with relapsing forms of multiple sclerosis (MS) and with a first clinical episode suggestive of MS treated up to 12 years. This review is an update to a previous version that examined data from the teriflunomide core clinical development program and extension studies. Data have since become available from active comparator trials with other disease-modifying therapies, treatment-related changes in brain volume (analyzed using structural image evaluation using normalization of atrophy) and real-world evidence including patient-reported outcomes. Initial data on the potential antiviral effects of teriflunomide in patients with MS, including case reports of patients infected with the 2019 novel coronavirus (SARS-CoV-2), are also presented.

    Lay abstract

    Teriflunomide, a treatment taken orally once a day, has shown consistent effectiveness and safety in patients with relapsing forms of multiple sclerosis (MS). This review is an update to a previous version that summarized the trials from when teriflunomide was in clinical development for MS. Some of the newer studies described here compared teriflunomide with other MS treatments. Studies have shown positive effects of teriflunomide on brain volume; teriflunomide may also be effective against some viruses. People taking teriflunomide generally report stable cognition and quality of life, with no worsening of fatigue or disability. In the EU, teriflunomide has been recently approved for use in pediatric patients 10 years of age and above.

    Practice points

    Dosage & administration

    • Teriflunomide is a once daily oral immunomodulator approved in USA (7 and 14 mg) since 2012 for the treatment of relapsing forms of multiple sclerosis (MS) and in the EU (14 mg) since 2013 for the treatment of relapsing-remitting MS.

    Mechanism of action: immunomodulation

    • Teriflunomide reversibly inhibits dihydroorotate dehydrogenase, a mitochondrial enzyme necessary for de novo pyrimidine synthesis (required for expansion of antigen-activated lymphocytes), and acts as an immunomodulator, reducing the activity of proliferating T cells and B cells and T helper type 1 cell counts and increasing CD4+/CD8+ T-cell ratios and regulatory T-cell counts.

    Pharmacodynamics, pharmacokinetics & metabolism

    • Oral bioavailability of teriflunomide is approximately 100%; pharmacokinetic characteristics are only minimally affected by food intake and mild-to-moderate hepatic or renal impairment, and elimination can be accelerated with an 11-day course of cholestyramine or activated charcoal.

    • Animal studies suggest teriflunomide is detectable in the blood, brain and cerebrospinal fluid, suggesting teriflunomide may have local activity in the central nervous system.

    Treatment efficacy

    • The clinical development program demonstrated the long-term efficacy of teriflunomide in adults with relapsing forms of MS.

    • The TERIKIDS trial demonstrated that times to first confirmed relapse and first disease activity were delayed with teriflunomide versus placebo in pediatric patients.

    • In clinical trials, teriflunomide is associated with reduced changes in brain volume, regardless of whether patients had disability worsening.

      Teriflunomide is associated with reductions in brain volume similar to ofatumumab, but less brain volume loss than with dimethyl fumarate.

    • In clinical trials, direct comparisons between teriflunomide and other disease-modifying therapies report varying results.

      In OPTIMUM, ponesimod was associated with lower annualized relapse rates and improvement in fatigue compared with teriflunomide.

      In ASCLEPIOS I and II, ofatumumab was associated with lower annualized relapse rates, fewer new magnetic resonance imaging lesions and lower rates of disability worsening compared with teriflunomide, but similar rates of disability improvement and brain volume loss.

      Patients switching from other disease-modifying therapies to teriflunomide report high overall patient treatment satisfaction, particularly in domains of convenience and treatment side effects.

    • Real-world observational studies using patient-reported outcomes report stable cognition, disability, fatigue and patient quality of life during teriflunomide treatment.

    Safety & tolerability

    • Teriflunomide has a well-characterized safety and tolerability profile that is consistent across studies and established with up to 12 years of exposure; recent data from observational and comparative effectiveness studies support the clinical trial data, with no new or unexpected safety signals.

    Multiple sclerosis (MS), an autoimmune demyelinating disease, is the leading cause of nontraumatic disability in young adults [1]. In both developed and developing countries, the incidence and prevalence of MS are increasing [1]. MS has an estimated prevalence of 1 in 3000 people; about 2.8 million people are estimated to live with the disease globally [2,3]. In Europe and the Americas, prevalence rates are 133 and 112, respectively, per 100,000 people [1,2]. Most patients present with relapsing-remitting MS (RRMS), characterized by periods of acute ‘relapses’ caused by inflammatory lesions, followed by periods of decreased activity or ‘remissions.’ Many patients with RRMS eventually transition to secondary progressive MS, which involves gradual worsening or accumulation of symptoms [3].

    Historically, key end points for most MS therapies have been annualized relapse rate (ARR), disability worsening and reduction of lesion numbers on magnetic resonance imaging (MRI) [3,4]. However, markers of acute focal inflammation do not correlate with long-term disability [5]. Although recent advances in disease-modifying therapies (DMT) have made it possible to effectively reduce the risk of focal inflammatory activity (and thus ARR), a need remains for improved therapeutic options that slow brain volume loss (BVL) and long-term disease progression. The lack of comprehensive head-to-head trials, real-world outcomes and patient-reported outcomes also drives a need for further research and understanding of MS treatments.

    A review of the DMT teriflunomide (AUBAGIO®, Sanofi Genzyme, MA, USA) published in 2017 summarized data from the core clinical development program and long-term extensions, real-world evidence and comparative effectiveness studies then available. This update to that review presents recently available evidence for teriflunomide, including direct comparisons with other DMTs in clinical studies, patient-reported outcomes, long-term effects on brain volume, evolving antiviral properties and outcomes for patients with MS treated with teriflunomide while infected with COVID-19.

    Overview of currently available disease-modifying therapies

    The first available DMTs for relapsing forms of MS were injectable interferons (IFNs: IFN-β-1a, IFN-β-1b, pegylated IFN-β-1a) [6–10] and glatiramer acetate (GA) [11], the earliest of which received US FDA approval in the 1990s (Table 1). In 2010 the first oral DMT, a sphingosine-1-phosphate (S1P) inhibitor (fingolimod), was approved to treat relapsing forms of MS [12]; approval of other S1P inhibitors followed in 2019 and 2020 [13,14]. Oral fumarates dimethyl fumarate (DMF), diroximel fumarate (DRF) and monomethyl fumarate (MMF) received approval in 2013, 2019 and 2020, respectively [15–17]. The purine antimetabolite cladribine was approved in 2019 [18]. Several monoclonal antibody (mAb) therapies have also been approved for use in MS. Natalizumab was the first mAb approved for this indication [19], followed by alemtuzumab [20], ocrelizumab [21] and ofatumumab [22].

    Table 1. Disease-modifying therapies approved by the US FDA for the treatment of multiple sclerosis.
    DMT categoryDMTIndicationRef.
    Injectable DMTsGlatiramer acetate (Copaxone®, Glatopa®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[11,84]
    IFN-β-1a (Rebif®, Avonex®)[8,9]
    IFN-β-1b (Betaseron®, Extavia®)[6,7]
    Mitoxantrone (Novantrone®)Treatment of progressive, progressive relapsing or worsening relapsing-remitting MS[85]
    Peginterferon beta-1a (Plegridy®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[10]
    Oral DMTsTeriflunomide (Aubagio®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[23]
    Sphingosine-1-phosphate inhibitors  
      Fingolimod (Gilenya®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[12]
      Ozanimod (Zeposia®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[13]
      Ponesimod (Ponvory™)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[86]
      Siponimod (Mayzent®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[14]
    Fumarates  
      Dimethyl fumarate (Tecfidera®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[15]
      Diroximel fumarate (Vumerity®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[16]
      Monomethyl fumarate (Bafiertam™)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[17]
    Purine antimetabolite  
      Cladribine (Mavenclad®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[18]
    Monoclonal antibodiesNatalizumab (Tysabri®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[19]
    Ocrelizumab (Ocrevus®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[21]
    Ofatumumab (Kesimpta®)Treatment of relapsing forms of MS, to include clinically isolated syndrome, relapsing-remitting disease and active secondary progressive disease, in adults[22]
    Alemtuzumab (Lemtrada®)§Treatment of relapsing forms of MS, to include relapsing-remitting disease and active secondary progressive disease, in adults[20]

    †Indicated for patients 10 years of age and older.

    ‡Natalizumab has a boxed warning regarding the increased risk of progressive multifocal leukoencephalopathy.

    §According to the US label, due to its safety profile, alemtuzumab should generally be reserved for patients with an inadequate response to two or more drugs indicated for the treatment of MS.

    DMT: Disease-modifying therapy; IFN: Interferon; MS: Multiple sclerosis.

    The precise mechanisms by which all DMTs act in patients with MS are unknown, and mechanism descriptions are beyond the scope of this article, but all effect reductions in ARR, rates of disability progression and/or new lesions on MRI. Some (e.g., alemtuzumab) have also been shown to significantly reduce brain atrophy [20].

    Teriflunomide

    The oral DMT teriflunomide was first approved by the US FDA in 2012 to treat relapsing forms of MS (Table 1) [23]. Teriflunomide is dosed once daily at 7 or 14 mg in USA but is available only at 14 mg in other countries [4]. In the EU, teriflunomide use was recently approved in pediatric patients 10 years of age and above, with a recommended dose dependent on body weight (>40 kg: 14 mg once daily; ≤40 kg: 7 mg once daily; stable body weight above 40 kg: should be switched to 14 mg once daily) [24]. The clinical development program, comprising the Phase III, randomized clinical trials TEMSO, TOWER, TENERE and TOPIC, demonstrated significant improvements in ARR, rates of disability worsening and MRI outcomes for teriflunomide versus placebo in patients with relapsing MS [23]. Teriflunomide has a robust effect on neurodegenerative processes, as reflected in the preservation of brain volume [25,26] and reduction of disability progression [27]. Clinical trial extension data, Phase IV studies and real-world evidence have demonstrated long-term safety, efficacy and tolerability. Compared with placebo, teriflunomide is associated with increased incidence of hepatic events (primarily reversible alanine aminotransferase [ALT] increases), lymphopenia and neutropenia (≤15% reduction, with lymphocyte and neutrophil counts remaining in the normal range) [4].

    Pharmacology

    Chemistry & mechanism of action

    The chemistry and proposed mechanism of action of teriflunomide were extensively covered in the 2017 publication of this review [4]. In brief, teriflunomide ([Z]-2-cyano-3-hydroxy-but-2-enoic acid-[4-trifluoromethylphenyl]-amide) is the principal active metabolite of leflunomide, and an open-ring malononitrile [4,23]. Leflunomide, the prodrug of teriflunomide, is rapidly converted in the intestinal mucosa or plasma, resulting in similar pharmacokinetics of the two agents [28]. Teriflunomide has a molecular mass of 270.2 Da, is practically insoluble in water (logP = 2.4), and has an acidic pKa of 5.48 [29].

    Teriflunomide is a reversible, selective inhibitor of dihydroorotate dehydrogenase (DHODH), a rate-limiting mitochondrial enzyme required for de novo pyrimidine synthesis, which is required for expansion of antigen-activated (but not resting) lymphocytes [28,29]. Inhibition of de novo pyrimidine synthesis leads to a reduction in activated T and B cells available to enter the central nervous system (CNS). Because resting lymphocytes do not require de novo pyrimidine synthesis for proliferation, teriflunomide only interrupts proliferation of activated lymphocytes without affecting the normal homeostatic process for resting lymphocytes [30]. Teriflunomide inhibits the binding to DHODH of dihydroorotate (in an allosteric, noncompetitive manner) and of the electron acceptor ubiquinone (in a competitive manner) [28]. Teriflunomide is not a nucleotide analog [30]. The Ames test shows it is not DNA intercalating and does not affect cell viability [30].

    In vitro and animal studies first suggested teriflunomide affects disease symptoms without inducing immunosuppression [30]. The effects of teriflunomide on size and function of human lymphocyte populations were explored in pooled analyses of the pivotal trials and separately in the Phase III, nonrandomized, open-label TERI-DYNAMIC study in patients with RRMS [31]. In TERI-DYNAMIC, lymphocyte and T helper type 1 cell (Th1) counts were reduced and CD4+/CD8+ T-cell ratios and induced regulatory T-cell (Treg) counts were increased after teriflunomide treatment [31]. In pooled analyses, absolute lymphocyte counts declined over 24 weeks of treatment (mean, 1.89 [baseline] to 1.67 [week 24]), then remained stable. Mean counts generally remained within the normal range, but grade 1 and 2 lymphopenia occurred in 7.3 and 2.2% of patients, respectively [32]. The occurrence of grade 2 lymphopenia was associated with decreased mean lymphocyte counts from baseline through week 48, after which lymphocyte counts stabilized [32]. Most cases of grade 1 or 2 lymphopenia occurred during the first year of treatment; fewer cases were reported after year 3, although the incidence of grade 1 lymphopenia rose slightly during years 6–8 [32].

    Lymphocyte and Th1 counts are reduced with teriflunomide, CD4+/CD8+ T-cell ratios and Treg counts are increased, and CD4+ clonal diversity is reduced. In TERI-DYNAMIC, following 24 weeks of treatment, total lymphocyte counts remained within the clinically normal range, although reductions from baseline were observed in absolute lymphocyte counts (-284.94; p < 0.001) and percentages of lymphocytes (-3.05%; p < 0.05), CD19+ B cells (-2.0%; p = 0.001) and CD8+ cells (-1.0%; p < 0.01), and increases occurred in CD4+ populations (1.6%; p < 0.01) and CD4+/CD8+ ratios [31]. In an analysis of CD4+ T-cell subpopulations, a selective effect on induced Treg, natural Treg and Th1 cells was seen, suggesting a shift in T-cell populations from proinflammatory to regulatory and potentially anti-inflammatory T cells [31]. Further, in vitro exposure to teriflunomide significantly reduced T-cell production of proinflammatory cytokines. In vitro and ex vivo findings from TERI-DYNAMIC supported a reversible effect of teriflunomide, and study results were consistent with observations from TEMSO and TOWER [4].

    In support of an immunomodulatory role, patients receiving teriflunomide are not at increased risk of malignancy or serious infection. Most teriflunomide-treated patients achieved seroprotection to both neoantigens and recall antigens in rabies and influenza vaccine studies, respectively. In a study using the rabies vaccine, all vaccinated patients achieved antibody titers >0.5 IU/ml and teriflunomide did not adversely affect the cellular memory response to recall antigens [33]. After receiving the 2011/2012 seasonal influenza vaccine in the Phase II TERIVA study, patients generally had an effective immune response, with >90% of patients achieving antibody titers ≥40 (considered protective immunity) for H1N1 and influenza B and 77–90% achieving titers ≥40 for H3N2 [34].

    Pharmacodynamics, pharmacokinetics & metabolism

    The pharmacokinetic properties of teriflunomide were summarized previously [4]. Briefly, teriflunomide exhibits 100% oral bioavailability and dose-dependent systemic exposure; pharmacokinetic characteristics are not significantly affected by food intake, mild or moderate hepatic impairment or severe renal impairment [23,28]. However, clearance rate is 23% lower in female than in male patients and teriflunomide is contraindicated in patients with severe hepatic impairment [23].

    Peak plasma levels occur 1–4 h after dosing, and elimination half-life is approximately 18–19 days for both dose levels. Elimination occurs primarily through direct biliary excretion of the unchanged drug (37.5% in the feces) and renal excretion (22.6% via urine) [23]. Teriflunomide undergoes extensive enterohepatic recirculation, resulting in very slow total plasma clearance [24]. Steady state is reached after approximately 3 months of treatment. However, this recycling also means cholestyramine or activated charcoal may be used to rapidly eliminate teriflunomide by >98% after 11 days [23]. Plasma protein binding is >99%, but volume of distribution is only 11 l [23]. Data suggest teriflunomide inhibits CYP2C8 and is a weak CYP1A2 inducer but does not affect pharmacokinetics of other CYP substrates, including bupropion (CYP2B6), midazolam (CYP3A4), S-warfarin (CYP2C9), omeprazole (CYP2C19) and metoprolol (CYP2D6) [23].

    A recent study using an experimental autoimmune encephalomyelitis (EAE) mouse model and fluorine-19 MRI detected teriflunomide in the blood (10.68–40.91 μg/g), brain (0.23–9.32 μg/g) and cerebrospinal fluid (0.74–2.68 μg/g) of healthy mice and EAE mice with brain lesions treated for over 13 days [35]. This finding supports a previous animal study that detected biologically relevant concentrations of teriflunomide in brain and spinal cord, suggesting teriflunomide may have local activity in the CNS [36].

    Clinical efficacy

    Clinical development program (Phase II & III studies)

    A Phase II study (NCT01487096) [37,38] and the Phase III TEMSO (NCT00134563) [27], TOWER (NCT00751881) [39], TOPIC (NCT00622700) [40] and TENERE (NCT00883337) [41] studies all demonstrated teriflunomide efficacy in adults with MS. Key outcomes of these trials and their extensions were described previously (Table 2) [4]. In these studies, patients treated with teriflunomide showed improvements over placebo in ARR, disability worsening and MRI outcomes, which were sustained in long-term extensions.

    Table 2. Summary of selected efficacy results from Phase II and Phase III clinical trials and their extension studies with teriflunomide.
    StudyStudy designPatient populationMedications comparedSelected relapse outcomesSelected MRI findingsAdditional efficacy outcomesRef.
    Phase II studies
    O'Connor et al. (2006)
    NCT01487096
    Randomized, double-blind, placebo-controlled; 36-weekRRMS; N = 179TER 7 mg
     TER 14 mg
     PBO
    Mean ± SD ARR:
     TER 7 mg: 0.58 ± 0.85
     TER 14 mg: 0.55 ± 1.12
     PBO: 0.81 ± 1.22
     (p = NS between groups)
    Median number of combined unique active lesions vs PBO:
     TER 7 mg: 0.2 (p < 0.03)
     TER 14 mg: 0.3 (p < 0.01)
    69% relative reduction in number of patients with EDSS (disability) increase at 36 weeks for TER 14 mg vs PBO (p < 0.04)[38]
    Kremenchutzky et al. (2016)
    NCT00228163
    Open-label extension; 13 yearsPatients completing
    Phase II trial and entering extension; N = 147
    TER 7 mg
     TER 14 mg
    Cumulative ARR:
     TER 7 mg: 0.26
     TER 14 mg: 0.19
    Reduction in mean number of newly Gd+ T1 lesions and newly active T2 lesions early in the extension; MRI activity remained low throughout (both doses)Mean EDSS scores remained stable throughout the extension (lower in TER 14 mg than in TER 7 mg)[37]
    Phase III studies
    O'Connor et al. (2011)
    TEMSO
     NCT00134563
    Randomized, double-blind, placebo-controlled; 108 weeksRelapsing MS; N = 1088TER 7 mg
     TER 14 mg
     PBO
    Adjusted ARR:
     TER 7 and 14 mg: 0.37 (p < 0.001 vs PBO)
     PBO: 0.54
     (31.5% reduction with TER 14 mg vs PBO)
    Relative reduction in total lesion volume vs PBO:
     TER 7 mg: 39.4% (p = 0.03)
     TER 14 mg: 67.4% (p < 0.001)
    Relative risk (vs PBO) of unique active lesions per scan, % (95% CI):
     TER 7 mg: 0.52 (0.42–0.65; p < 0.001)
     TER 14 mg: 0.31 (0.23–0.41; p < 0.001)
    Significantly fewer patients with sustained disability progression (≥12 weeks) for TER 14 mg (20.2%) vs PBO (27.3%; p = 0.03)[27]
    O'Connor et al. (2016)
    TEMSO extension
     NCT00803049
    9-year extensionPatients completing TEMSO and entering extension; N = 742TER 7 mg
     TER 14 mg
    ARRs among patients continuing treatment:
     TER 7 mg: 0.198
     TER 14 mg: 0.171
    ARRs in patients randomized from PBO to TER:
     TER 7 mg: 0.225
     TER 14 mg: 0.177
    Reduction in mean number of Gd+ T1 lesions after patients switched from PBO to TERMean EDSS scores remained stable throughout the extension[87]
    Confavreux et al. (2014)
    TOWER
     NCT00751881
    International, randomized, double-blind, placebo-controlled; treatment ≤48 weeks after enrollment of last patientRelapsing MS; N = 1169TER 7 mg
     TER 14 mg
     PBO
    ARR (95% CI):
     TER 7 mg: 0.39 (0.33–0.46)
     TER 14 mg: 0.32 (0.27–0.38)
     PBO: 0.50 (0.43–0.58)
    NRTER 14 mg reduced the risk of sustained accumulation of disability vs PBO (HR, 0.68; 95% CI, 0.47–1.00; p = 0.044)
    LS mean change in EDSS score:
     TER 7 mg: 0.04 (p = NS vs PBO)
     TER 14 mg: -0.05 (p = 0.043 vs PBO)
     PBO: 0.09
    [39]
    Miller et al. (2020)
    TOWER extension
     NCT00751881
    6.3-year extensionPatients completing TOWER and entering extension; N = 751TER 14 mgCumulative adjusted ARRs through core and extension:
     Core PBO/extension TER 14 mg: 0.3
     Core TER 7 mg/extension TER 14 mg: 0.3
     TER 14 mg/TER 14 mg: 0.2 (p = 0.032 vs PBO/TER 14 mg)
    NRChanges in EDSS through week 96 were similar between groups
     Compared with PBO/TER 14 mg group, time to 12-week CDW was 12.3% lower and 23.9% for patients receiving TER 7 mg/TER 14 mg vs TER 14 mg/TER 14 mg
    [88]
    Miller et al. (2014)
    TOPIC
     NCT00622700
    Randomized, double-blind, placebo-controlled; 108 weeksPatients with first clinical episode suggestive of MS; N = 618TER 7 mg
     TER 14 mg
     PBO
    Risk of relapse determining conversion to CDMS vs PBO, HR (95% CI):
     TER 7 mg: 0.628 (0.416–0.949; p = 0.027)
     TER 14 mg: 0.574 (0.379–0.869; p = 0.009)
    Adjusted ARRs:
     TER 7 mg: 0.190
     TER 14 mg: 0.194
     PBO: 0.284
    Patients with new MRI lesions:
     TER 7 mg: 54%
     TER 14 mg: 52%
     PBO: 62%
    Patients with 12-week CDW:
     TER 7 mg: 10%
     TER 14 mg: 7%
     PBO: 10%
    EDSS score change from baseline, LS mean ± SE difference vs PBO:
     TER 7 mg: -0.259 ± 0.121 (p = 0.033)
     TER 14 mg: -0.235 ± 0.116 (p = 0.044)
    [40]
    Miller et al. (2019)
    TOPIC extension
     NCT00622700
    5.5-year extensionPatients completing TOPIC and entering extension; N = 423TER 7 mg
     TER 14 mg
    Risk of relapse determining conversion to CDMS, HR (95% CI):
     Core TER 7 mg/extension TER 7 mg: 1.560 (0.886–2.747); p = 0.123 vs Core PBO/extension TER 7 mg
     Core TER 14 mg/extension TER 14 mg: 0.529 (0.317–0.883); p = 0.015 vs Core PBO/extension TER 14 mg
     Core TER 14 mg/extension TER 14 mg: 0.604 (0.394–0.925); p = 0.021 vs Core TER 7 mg/extension TER 7 mg
     Adjusted ARR across treatment groups: ≤0.150
    Reductions from baseline in number of T1 Gd+ lesions per scan at week 204, mean (SD):
     Core PBO/extension TER 7 mg: 0.200 (0.422)
     Core TER 7 mg/extension TER 7 mg: 0.188 (2.070)
     Core PBO/extension TER 14 mg: -1.000 (2.256)
     Core TER 14 mg/extension TER 14 mg: -1.636 (5.320)
    Patients free of 12-week CDW:
     Core PBO/extension TER 7 mg: 84.4%
     Core TER 7 mg/extension TER 7 mg: 74.7%
     Core PBO/extension TER 14 mg: 77.0%
     Core TER 14 mg/extension TER 14 mg: 80.6%
    EDSS score change from baseline at week 204, mean (SD):
     Core PBO/extension TER 7 mg: -0.140 (0.729)
     Core TER 7 mg/extension TER 7 mg: 0.043 (1.023)
     Core PBO/extension TER 14 mg: -0.058 (1.105)
     Core TER 14 mg/extension TER 14 mg: -0.180 (0.925)
    [89]
    Chitnis (2020)
     TERIKIDS
     NCT02201108
    Randomized, double-blind, placebo-controlled; 96 weeksChildren (age 10–17 years) with RRMS; N = 166TER 7–14 mg (adult equivalent)
     PBO
    Median time to clinical relapse:
     TER: 75.3 weeks
     PBO: 39.1 weeks (34% relative risk reduction; p = 0.29)
    Sensitivity analysis of median time to relapse or high MRI activity:
     TER: 72.1 weeks
     PBO: 37.0 weeks
     (43% relative risk reduction; p = 0.04)
    Number of T1 Gd+ lesions:
     TER: 1.9
     PBO: 7.5
     (75% relative reduction; p < 0.0001)
    Number of new/enlarging T2 lesions:
     TER: 4.7
     PBO: 10.5
     (55% relative reduction; p < 0.001)
    % change in T2 lesion volume, TER vs PBO:
     Week 24: 0.08 vs 0.15%; p = 0.046
     Week 72: 0.07 vs 0.20%; p = 0.005
     Week 96: 0.07 vs 0.20%; p = 0.01
    NR[42]

    AE: Adverse event; ARR: Annualized relapse rate; CDMS: Clinically definite multiple sclerosis; CDW: Confirmed disability worsening; EDSS: Expanded Disability Status Scale; Gd+: Gadolinium-enhancing; HR: Hazard ratio; LS: Least squares; MRI: Magnetic resonance imaging; NR: Not reported; NS: Not significant; PBO: Placebo; TER: Teriflunomide.

    The TERIKIDS study evaluated time to first relapse and MRI outcomes with teriflunomide in 166 children (10–17 years old) with relapsing MS. The study, which has been presented in abstract form [42,43], included 96 weeks of double-blind, randomized, placebo-controlled treatment with 14 mg adult equivalent of teriflunomide or placebo, followed by an open-label extension with up to 192 weeks of teriflunomide. In addition to the primary end point of time to first clinical relapse, the study also included a prespecified sensitivity analysis of time to first relapse or high MRI activity. While teriflunomide reduced the risk of clinical relapse by 34% compared with placebo (median time to relapse, 75.3 vs 39.1 weeks, respectively), the difference was not statistically significant (p = 0.29). This is possibly because more patients than anticipated switched from the double-blind to open-label periods due to high MRI activity, which is consistent with clinical practice where relapses and MRI findings are routinely integrated in the clinical management of pediatric MS patients, including treatment selection. Switches were more frequent and occurred earlier in the placebo group (teriflunomide, 13%; placebo, 26%), leading to reduced study power and potentially biasing results against treatment efficacy. The prespecified sensitivity analysis detected statistically significantly longer median times to clinical relapse or switch due to high MRI activity with teriflunomide (72.1 weeks) compared with placebo (37.0 weeks; 43% relative risk reduction; p = 0.04). Statistically significant reductions were also observed in the adjusted number of T1 gadolinium-enhancing (Gd+) lesions per MRI scan (teriflunomide, 1.9; placebo, 7.5; 75% relative reduction; p < 0.0001) and T2 lesions (4.7 vs 10.5, respectively; 55% relative reduction; p = 0.0006) and least-squares mean change from baseline in T2 lesion volume at weeks 24 (0.08 vs 0.15; p = 0.046), 72 (0.07 vs 0.20; p = 0.005) and 96 (0.07 vs 0.20; p = 0.01) [42].

    Comparative effectiveness

    Teriflunomide has been used as an active comparator in recent Phase III studies (Table 3). The previous review described the TENERE study and extension, which compared teriflunomide 7 and 14 mg with IFN-β-1a 44 μg [4,41]. A 2018 analysis of patient-reported outcomes from the 96-week TENERE extension described statistically significant (p < 0.01) improvements in patient satisfaction related to side effects and convenience after switching from IFN-β-1a to teriflunomide [44]. The OPTIMUM study randomized patients with relapsing MS to ponesimod 20 mg once daily (QD) or teriflunomide 14 mg QD. At week 108, the ponesimod group had a significantly lower ARR compared with teriflunomide (0.202 vs 0.290; p = 0.0003) and significantly improved fatigue scores [45]. ASCLEPIOS I and II compared teriflunomide 14 mg QD with ofatumumab 20 mg every 4 weeks, reporting lower ARRs, fewer Gd+ T1 and new/enlarging T2 lesions, and lower rates of disability worsening with ofatumumab versus teriflunomide, but similar rates of clinical disability improvement and BVL reduction [46].

    Table 3. Summary of selected efficacy results from clinical trials of comparative effectiveness with teriflunomide.
    StudyStudy designPatient populationMedications comparedSelected relapse outcomesSelected MRI findingsDiscontinuation ratesOther efficacy outcomesRef.
    Kappos et al. (2019)
    OPTIMUM
     NCT0245644
    Double-blind, Phase IIIRelapsing MS; N = 1133TER 14 mg QD
     Ponesimod 20 mg QD
    Reductions in ARRs at week 108 for ponesimod vs TER: 0.202 vs 0.290 (rate ratio, 0.695; 99% CL, 0.536–0.902; p = 0.0003)NRNRFatigue significantly improved for ponesimod vs TER at week 108 (FSIQ-RMS mean difference, -3.57; p = 0.0019)
     No treatment differences in confirmed disability accumulation at weeks 12 or 24
    [45]
    Vermersch et al. (2018)
    TENERE
     NCT00883337
    TENERE; randomized, rater-blinded, Phase IIIRelapsing MS; N = 324TER 7 or 14 mg
     IFN-β-1a 44 μg
    Adjusted ARR:
     TER 7 mg: 0.41 (p = 0.03 vs IFN-β-1a)
     TER 14 mg: 0.26 (p = NS vs IFN-β-1a)
     IFN-β-1a: 0.22
    NRDiscontinuation rates:
     TER 7 mg: 6.4%
     TER 14 mg: 13.5%
     IFN-β-1a: 24.0% (18.3% due to AEs)
    Cumulative percentage of estimated treatment failure:
     TER 7 mg: 36%
     TER 14 mg: 33%
     IFN-β-1a: 37%
    [44]
    Hauser et al. (2020)
    ASCLEPIOS I and II
     NCT02792218
     NCT02792231
    Double-blind, double-dummy, Phase IIIRelapsing MS; ASCLEPIOS I, N = 927
     ASCLEPIOS II, N = 955
    TER 14 mg QD
     OFA 20 mg every 4 weeks
    ARRs, TER vs OFA:
     ASCLEPIOS I: 0.22 vs 0.11 (p < 0.001)
     ASCLEPIOS II: 0.25 vs 0.10 (p < 0.001)
    Mean number of Gd+ T1 lesions, TER vs OFA:
     ASCLEPIOS I: 0.45 vs 0.01; rate ratio, 0.03; 95% CI, 0.01–0.05; p < 0.001
     ASCLEPIOS II: 0.51 vs 0.03; rate ratio, 0.06; 95% CI, 0.04–0.10; p < 0.001
     Mean number of new/enlarging T2 lesions/year, TER vs OFA:
     ASCLEPIOS I: 4.00 vs 0.72; rate ratio, 0.18; 95% CI, 0.15–0.22; p < 0.001
     ASCLEPIOS II: 4.15 vs 0.64; rate ratio, 0.15; 95% CI, 0.13–0.19; p < 0.001
    Across trials and treatment groups, AEs led to discontinuation for 5.2–5.8% of patientsRates of CDW (pooled analyses), TER vs OFA:
     3-month: 15.0 vs 10.9%; HR, 0.66; 95% CI, 0.50–0.86; p = 0.002
     6-month: 12.0 vs 8.1%; HR, 0.68; 95% CI, 0.50–0.92; p = 0.01
    Rates of 6-month clinical disability improvement, TER vs OFA: 8.1 vs 11.0%; HR, 1.35; 95% CI, 0.95–1.92; p = 0.09
    [46]

    AE: Adverse event; ARR: Annualized relapse rate; CDW: Confirmed disability worsening; CI: Confidence interval; CL: Confidence limit; FSIQ-RMS: Fatigue Symptoms and Impacts Questionnaire-Relapsing Multiple Sclerosis; Gd+: Gadolinium-enhancing; HR: Hazard ratio; IFN: Interferon; MS: Multiple sclerosis; NR: Not reported; NS: Not significant; OFA: Ofatumumab; QD: Once daily; TER: Teriflunomide.

    An analysis using data from pivotal studies of teriflunomide (TEMSO and TOWER), DMF (DEFINE and CONFIRM) and fingolimod (FREEDOMS and FREEDOMS II) calculated numbers needed to treat (NNT) to prevent relapses or disability worsening. NNTs were generally comparable for the prevention of any relapse (5.6–5.9 vs 5.3–5.6 vs 4.5–5.3, respectively), relapses leading to hospitalization (12.2–20.0 vs 50.0–58.8 vs 9.1–83.3) or requiring intravenous corticosteroids (6.3–6.9 vs 6.3–6.7 vs 4.8–6.9), and disability worsening (13.7–17.1 vs 10.8–30.2 vs 15.3–23.5). However, differences in study design and patient populations should be considered when evaluating cross-trial comparisons [47].

    Real-world evidence

    Observational studies have assessed real-world outcomes for patients with relapsing forms of MS treated with teriflunomide. The Phase IV Teri-PRO study was described in the previous review [4], but additional results have since been reported [48,49]. Teri-PRO (N = 1000) used patient-reported outcomes to assess treatment satisfaction and changes in disability, cognition and quality of life over 48 weeks. Such assessments are important to help clinicians understand the impact of treatment on patients' daily life, which may not be captured in traditional clinical study end points. Patients reported high overall treatment satisfaction on the Treatment Satisfaction Questionnaire for Medication scale (TSQM; mean score, 68.2) [49]. Patients who had switched from another DMT to teriflunomide reported significantly improved satisfaction (p < 0.001) at week 48 compared with baseline, both overall and across TSQM subscales of effectiveness, convenience and side effects (Figures 1 & 2) [48]. In the overall population, the highest scores were in the side effects (84.1) and convenience (90.4) TSQM subdomains. These subdomains corresponded with the most common reasons for switching to teriflunomide (i.e., convenience of an oral formula and safety concerns around other DMTs) [49]. Disability scores remained stable throughout the study, including patient-reported disability, as measured by the Patient-Determined Disease Steps (PDDS) scale, and Expanded Disability Status Scale (EDSS) scores [49]. Quality-of-life scores measured using the Multiple Sclerosis International Quality of Life (MusiQoL) significantly improved from baseline to week 48 (mean [95% CI], 67.7 [66.7–68.6] to 69.2 [68.1–70.2]; p = 0.003), with most subscale scores remaining stable or improving over time. Similarly, scores remained stable for the Stern Leisure Activity Scale (mean [95% CI], 7.30 [7.16–7.44] to 7.40 [7.24–7.56]) and two scales measuring cognition. The annualized treated relapse rate was 0.20 [49].

    Figure 1. Distribution of patients by degree of disability, as measured by MSPS subscales, at baseline and week 48 in all patients in the real-world Teri-PRO study.

    Normal/minimal, mild/moderate and severe/total categories refer to MSPS scores of 0/1, 2/3 and 4/5, respectively, except for the Mobility subscale, where MSPS scores for each category were 0/1, 2/3/4, and 5/6, respectively.

    BL: Baseline; MSPS: Multiple Sclerosis Performance Scale; W: Week.

    Reproduced from [49].

    Figure 2. Distribution of patients by degree of disability, as measured by MSPS subscales, at baseline and week 48 in patients switching to teriflunomide from another DMT in the real-world Teri-PRO study.

    Normal/minimal, mild/moderate and severe/total categories refer to MSPS scores of 0/1, 2/3 and 4/5, respectively, except for the Mobility subscale, where MSPS scores for each category were 0/1, 2/3/4 and 5/6, respectively.

    BL: Baseline; DMT: Disease-modifying therapy; MSPS: Multiple Sclerosis Performance Scale; W: Week.

    Reproduced from [48].

    The observational TACO study (N = 47) assessed patient-reported and clinical outcomes after 24 months of teriflunomide treatment [50]. The ARR remained low at 24 months (0.14; 95% CI, 0.07–0.25), EDSS scores remained stable compared with baseline and 80.9% of patients did not experience disability worsening. Assessments of quality of life via the Multiple Sclerosis Impact Scale (MSIS-29) suggested stable physical and psychologic status, with mean (95% CI) changes from baseline to month 24 of 0.0 (-5.4 to 5.5; p = 0.986) and -3.6 (-8.9 to 1.6; p = 0.374), respectively. Treatment satisfaction, effectiveness, side effects, and convenience scores improved, with mean (95% CI) changes from baseline to month 24 of 15.4 (-1.2 to 32.1), 4.5 (-10.4 to 19.4), 22.8 (1.2–44.4), and 21.2 (10.3–32.2), respectively. Cognition, anxiety/depression, and fatigue scores remained relatively stable over 24 months, with small or no changes from baseline [50].

    TAURUS-MS was a prospective observational study of 1128 patients with RRMS, for whom mean ARR decreased from 0.87 in the 2 years before study entry to 0.35 during the 2-year observational period (p ≤ 0.001) [51]. EDSS and fatigue scores remained stable throughout treatment. TSQM scores improved from baseline to 24 months among patients who received another DMT before study entry (global satisfaction, 15.3-point improvement; effectiveness, 8.1; convenience, 17.0; p < 0.001 for all).

    Patient-reported outcomes were also evaluated in the observational Teri-FAST (N = 210) [52], Teri-REAL (N = 100) [53] and Teri-LIFE (N = 200) [54] studies. Teri-FAST assessed fatigue (via EMIF-SEP, a version of the Modified Fatigue Impact Scale [MFIS]), depression (Beck Depression Inventory [BDI]) and health-related quality of life (Two-Life Scale [TLS-QoL 10]). Between baseline and year 2, patients reported stable fatigue (mean change, -1.54; 95% CI -4.02 to 0.94), depression (-0.6; -1.5 to 0.3) and health-related quality of life (-0.3; -0.8 to 0.1) [52]. Similarly, physical and psychologic domains of the MSIS-29 remained stable throughout Teri-REAL (mean ± SD changes from baseline to month 12, 2.4 ± 16.5 [p = 0.265] and 0.0 ± 20.7 [p = 0.990], respectively) [53]. Treatment satisfaction measured using TSQM remained steady or improved (global satisfaction, 0.9 ± 3.4 [p = 0.075]; convenience, 1.1 ± 2.6 [p = 0.006]; effectiveness, 0.5 ± 4.6 [p = 0.480]). Teri-LIFE reported relatively stable physical and mental quality of life measured via the Short Form-36 (SF-36), with mean (95% CI) 24-month changes from baseline of -1.5 (-2.8 to -0.2; p = 0.0038) and 1.6 (-0.2 to 3.5; p = 0.115), respectively. In Teri-LIFE the on-treatment ARR was 0.17 (95% CI, 0.13–0.23) [54].

    Safety & tolerability

    The safety and tolerability profile of teriflunomide is well established and consistent across studies, including in clinical trial and real-world settings. The previous review summarized the overall safety data for teriflunomide, including results from a pooled analysis of Phase II and Phase III pivotal studies. In that analysis, adverse events (AEs) were similar between the core studies and the study extensions, with no new or unexpected events occurring during longer-term use in extension studies [4,55].

    Real-world evidence supports the safety profile seen in clinical trials. About 82% of patients in Teri-PRO experienced AEs, most of which were of mild or moderate severity and included hair thinning, diarrhea, nausea, headache, urinary tract infection and ALT increase (Table 4) [48,49]. AEs led to treatment discontinuation for 10.9% of patients. MS relapse was the only serious AE reported for >1% of patients, and diarrhea and MS relapse were the only AEs leading to discontinuation in >1% of patients. In TACO, 21.8% of patients experienced treatment-related AEs, most commonly hair thinning (10.9%) and diarrhea (9.1%), and none reported serious AEs [50]. TAURUS-MS reported AEs for 35.8% of patients, the most frequent of which were diarrhea, MS relapse, hair thinning and viral upper respiratory tract infection (Table 5). Serious AEs in TAURUS-MS included one death due to bronchopulmonary aspergillosis that was assessed by the investigator as related to teriflunomide, although confounding factors may have played a role [51].

    Table 4. Adverse events reported in the Teri-PRO study.
    AEPatients, n (%)
    Patients with ≥1 AE823 (82.3)
    AEs reported in ≥5% of patients
      Hair thinning230 (23.0)
      Diarrhea173 (17.3)
      Nausea82 (8.2)
      Headache69 (6.9)
      Urinary tract infection67 (6.7)
      ALT increase63 (6.3)
      Nasopharyngitis54 (5.4)
      Fatigue52 (5.2)
    Patients with ≥1 serious AE127 (12.7)
    Serious AEs reported in ≥5 patients
      MS relapse21 (2.1)
      Hypertension6 (0.6)
      ALT increase6 (0.6)
      Urinary tract infection5 (0.5)
    Patients with permanent treatment discontinuation due to an AE109 (10.9)
    AEs leading to permanent treatment discontinuation in ≥5 patients
      Diarrhea17 (1.7)
      MS relapse12 (1.2)
      Hair thinning9 (0.9)
      ALT increase6 (0.6)
    Patients with AE leading to death4 (0.4)

    N = 1000.

    †Listed by MedDRA-preferred term.

    ‡MedDRA-preferred term is alopecia.

    AE: Adverse event; ALT: Alanine aminotransferase; MedDRA: Medical Dictionary for Regulatory Activities (Version 18.1); MS: Multiple sclerosis.

    Data taken from [49].

    Table 5. Adverse events with incidence ≥1% in the observational TAURUS-MS study.
    AEPatients with AE, n (%) (N = 1139)
    Patients with any AE408 (35.8)
    Serious AEs148 (13.0)
    Diarrhea55 (4.8)
    MS relapse48 (4.2)
    Hair thinning38 (3.3)
    Viral upper respiratory tract infection31 (2.7)
    Influenza22 (1.9)
    Drug ineffective19 (1.5)
    Urinary tract infection18 (1.6)
    Bronchitis17 (1.5)
    Hypertension16 (1.4)
    Influenza-like illness16 (1.4)
    Nausea15 (1.3)

    †Medical Dictionary for Regulatory Activities (MedDRA) preferred term is alopecia.

    AE: Adverse event; MedDRA: Medical Dictionary for Regulatory Activities (Version 18.1); MS: Multiple sclerosis.

    Adapted from [51].

    Recent publications of comparative studies with teriflunomide now permit safety comparisons between teriflunomide and other treatments. A pooled analysis of ASCLEPIOS I and II reported the rates of AEs for teriflunomide and ofatumumab were 84.2 and 83.6%, respectively, and rates of serious AEs were 7.9 and 9.1%, respectively [46]. In that analysis, infections and serious infections were reported for 52.7 and 1.8% of patients who received teriflunomide and 51.6 and 2.5% who received ofatumumab. Injection-related systemic reactions occurred in 15.0% of patients in the teriflunomide group who received placebo injections and 20.2% in the ofatumumab group. One of the two (0.2%) grade 3 injection-related systemic reactions in the ofatumumab group led to treatment discontinuation. Malignancies were reported for four (0.4%) patients receiving teriflunomide and five (0.5%) receiving ofatumumab [46].

    In TERIKIDS, children receiving teriflunomide had higher infection rates (66.1%; 1.38 events/person-year) compared with those receiving placebo (45.6%; 0.99 events/person-year) [42]. However, rates of any AEs and serious AEs were similar between the teriflunomide and placebo groups (any AEs, 88.1 vs 82.5%, respectively; serious AEs, 11.0 vs 10.5%). AEs occurring with high frequency with teriflunomide (≥5% more than placebo) included nasopharyngitis, hair thinning, upper respiratory tract infection, paresthesia, abdominal pain and increased blood creatine phosphokinase. Two teriflunomide-treated patients experienced acute pancreatitis during the double-blind period, leading to treatment discontinuation and one of these events led to hospitalization and required corrective treatment [24,42]. No deaths were observed in the placebo or teriflunomide groups. Overall, the beneficial effects of teriflunomide were similar to those seen in adults, with a manageable safety profile.

    To date, teriflunomide has not been associated with QT prolongation [23], but the US label does have a boxed warning regarding the risk of hepatotoxicity and embryofetal toxicity [23]. Given these risks, liver monitoring should be done within 6 months before treatment initiation and monthly for 6 months after treatment initiation. Teriflunomide is associated with a 10% reduction in platelet count and a 15% reduction in white blood cell count (primarily neutrophils and lymphocytes) during the first 6 weeks, which remained low throughout treatment [23].

    Teriflunomide is contraindicated in pregnancy based on data from preclinical animal studies, but data in human pregnancy are limited. Patients not wishing to become pregnant should use reliable contraception. Patients wishing to become pregnant may undergo an 11-day accelerated elimination program [23]. The teriflunomide clinical development program required patients to use reliable contraception and prohibited pregnant women from participating. The prescribing information for teriflunomide also recommends that women of childbearing potential use reliable contraception during treatment and during an accelerated elimination procedure (if treatment is discontinued). Despite these precautions, more than 400 pregnancies (222 with known outcomes) were reported by December 2017 in clinical trials and real-world use [56]. The previous review described the 70 pregnancies reported for women in the clinical development program [4]. In this update, all pregnancies with known outcomes are described in Table 6.

    Table 6. Outcomes of teriflunomide-exposed pregnancies.
    Patients Pregnancy outcome, n (%)Birth defects in live births or stillbirths, nRef.
      Live birthStillbirthSpontaneous abortionElective abortionEctopic pregnancyFetal death secondary to maternal deathMajor birth defectMinor birth defect 
      All live birthsPreterm live birth (<37 weeks)
    Female patients exposed to teriflunomideClinical study (n = 70)27 (38.6)2/27 (7.4)010 (14.3)31 (44.3)2 (2.9)01: ureteropyeloectasia0[56]
    Prospective/postmarketing
    (n = 91)
    49 (53.8)4/49 (8.2)021 (23.1)19 (20.9)#1 (1.1)1 (1.1)03: congenital hydrocephalus, ventricular septal defect, malformation of right foot valgus
    Retrospective/postmarketing (n = 61)31 (50.8)3/31 (9.7)1 (1.6)§16 (26.2)13 (21.3)0000
    Overall (n = 222)107 (48.2)9/107 (8.4)1 (0.5)47 (21.2)63 (28.4)3 (1.4)1 (0.5)13
    Female partners of male patients exposed to teriflunomideProspective (n = 23)20 (87.0)002 (8.7)1 (4.3)0001: congenital pes planus[57]
    Retrospective (n = 25)17 (68.0)03 (12.0)4 (16.0)1 (4.0)0001: congenital hip dysplasia
    Overall (n = 48)37 (77.1)03 (6.3)6 (12.5)2 (4.2)0002
    Female partners of male patients exposed to leflunomide††Prospective (n = 37)31 (83.8)1 (2.7)04 (10.8)1 (2.7)1 (2.7)000[57]
    Retrospective (n = 20)6 (30.0)2 (10.0)2 (10.0)5 (25.0)7 (35.0)‡‡001: bilateral renal agenesis0
    Overall (n = 57)37 (64.9)3 (5.3)2 (3.5)9 (15.8)8 (14.0)1 (1.8)010

    †107 live births including three sets of twins and one set of triplets (112 babies in total).

    ‡Reported as percent of live births.

    §Fetal death occurred at 35 weeks. Cause and details are unknown.

    ¶Includes one set of twins.

    #Including one elective termination following prenatal diagnosis of blighted ovum.

    ††Leflunomide is the parent drug of teriflunomide.

    ‡‡Termination was for fetal anomaly.

    The only major birth defect associated with teriflunomide exposure was a case of ureteropyeloectasia. Five minor birth defects were reported in live births or stillbirths (congenital hydrocephalus, ventricular septal defect, malformation of right foot valgus, congenital pes planus, congenital hip dysplasia) [56,57]. In the one major and three minor birth defects in infants born to women receiving teriflunomide, teriflunomide exposure occurred only during the first trimester [56]. In a study of 48 pregnancies in female partners of men treated with teriflunomide, no detectable embryofetal toxicity signal was observed [57]. The rates of major and minor birth defects and spontaneous abortions in the two analyses were considered comparable to, or below, those in the general population [56,57]. These findings are encouraging for patients who unknowingly take teriflunomide in the early stages of pregnancy, but further data are needed before conclusions can be drawn regarding treatment safety in pregnancy.

    A population-based analysis of pregnancy data from the Danish Multiple Sclerosis Registry described 31 pregnancies in 13 women receiving teriflunomide and 18 partners of teriflunomide-treated men. Female patients with MS had 11 elective abortions and two live births, neither with congenital malformations. All 18 pregnancies in female partners of treated men led to live births, with one congenital malformation reported (plagiocephaly). Compared with controls from the general population, teriflunomide-exposed pregnancies were associated with 22% reduced odds of any AE, although the association did not reach statistical significance (95% CI, 0.16–3.72; p = 0.753) [58].

    Antiviral effects

    Some evidence suggests MS results from a combination of genetic and environmental factors [59], and exposure to certain viruses may play a critical role in the development of MS and occurrence of relapses [60,61]. Primary exposure to, and reactivation of, some viruses may trigger or worsen disease progression and increase exacerbation risk [62]. In addition, the presence of certain viruses, such as John Cunningham virus (JCV), increases the risk of serious complications (e.g., progressive multifocal leukoencephalopathy [PML]) [63]. DMTs have varying effects on immunomodulation or immunosuppression, and long-term use of some DMTs may further increase patients' susceptibility to viral infection [64].

    Preclinical evidence suggests teriflunomide reduces production or viral loads of herpes simplex virus-1 and the BK polyomavirus, which is closely related to JCV [65]. In vitro studies also suggest teriflunomide may inhibit JCV infection and spread in a dose-dependent manner [66]. No medically confirmed cases of PML matching the American Academy of Neurology (AAN) criteria have been attributed to teriflunomide to date, with over 435,246 patient-years of teriflunomide exposure in MS [67,68], whereas PML has been reported in patients treated with DMF, fingolimod and natalizumab [64].

    The potential antiviral properties of teriflunomide were further supported by an ex vivo study that suggested teriflunomide inhibits abnormal T-cell proliferation associated with human T-lymphotrophic virus-1 (HTLV-1) infection, which causes HTLV-1–associated myelopathy/tropical spastic paraparesis, a chronic inflammatory CNS disease [69].

    Based on in vitro studies suggesting teriflunomide may have antiviral activity against Epstein-Barr virus (EBV), a clinical study was conducted in 19 patients receiving teriflunomide 14 mg to treat RRMS [70]. The study used reference cohorts comprising 157 patients with MS (untreated or receiving another DMT) who had salivary samples assessed for the presence of EBV DNA. The proportions of patients with salivary EBV DNA or EBV shedding were numerically lower for teriflunomide versus the reference cohorts, and significantly fewer salivary samples from teriflunomide-treated patients were positive for EBV DNA or EBV shedding. Of the 11 teriflunomide-treated patients positive for EBV DNA, two patients accounted for the majority of EBV shedding [70]. The study provided preliminary evidence suggesting the possibility teriflunomide may have antiviral effects on EBV load. A separate study in 30 patients with MS reported reductions in EBV nuclear antigen-1 and viral capsid antigen titers compared with healthy controls during the first 12 months of teriflunomide treatment [71]. Patients with the greatest reductions from baseline had significantly less cortical and gray matter BVL.

    As described above, studies of rabies and influenza vaccines in patients with MS suggest most teriflunomide-treated patients achieved seroprotection to neoantigens and recall antigens, respectively [33,34].

    Experience of COVID-19 in teriflunomide-treated patients

    This article was prepared during the global outbreak of the 2019 novel coronavirus (SARS-CoV-2) that causes COVID-19. Given the varying effects of DMTs on immunomodulation and immunosuppression, concerns have arisen that DMTs might increase patient risk of SARS-CoV-2 infection, impair the immune response, or contribute to poor outcomes [72]. The influence (if any) of teriflunomide treatment on the course of COVID-19 has therefore been of concern to physicians and patients. One case series described five patients with MS treated with teriflunomide who were infected with SARS-CoV-2 [72]. Patients included three men and two women aged 38–79 years with EDSS scores of 0–6 who had received teriflunomide 14 mg/day for up to 4 years. Three had comorbidities, and two had a body mass index >25 kg/m2 (28.5 and 29.9 kg/m2). All five continued a stable teriflunomide dose during infection and had self-limiting infection that did not require hospitalization or lead to MS relapse. In all cases, symptoms resolved ≤39 days after COVID-19 diagnosis [72].

    A separate case series reported that six patients with MS continued teriflunomide treatment without experiencing MS relapse during symptomatic COVID-19 infection [73]. Patients included four women and two men aged 34–57 years with EDSS scores of 1–4.5 who had been receiving teriflunomide for 0.5–5 years. None had relevant comorbidities or required hospitalization during COVID-19 infection, and all recovered without receiving specific treatment. One patient reported diplopia, which was interpreted by the treating physician as a pseudorelapse [73].

    In a separate report, a 42-year-old man receiving teriflunomide for RRMS was hospitalized for 4 days and treated with high-dose methylprednisolone for neurologic symptoms presumed to indicate MS relapse. The day after discharge, neurologic symptoms deteriorated and the patient developed fever, hypotension, tachycardia and increased serum acute phase reactant (APR) levels, leading to rehospitalization and COVID-19 diagnosis. The patient received symptomatic treatment (no supplemental oxygen) and was discharged 1 week later, following resolution of fever and reduction of APR levels [74]. The authors did not report whether teriflunomide was continued during the second hospitalization.

    In a 62-year-old woman with RRMS (EDSS score = 6), teriflunomide was discontinued during a 6-day hospitalization for COVID-19–related pneumonia, but plasma levels of teriflunomide remained >10 mg/l at discharge. Based on analysis of CD4+ and CD8+ T cells before and during infection, the authors suggested teriflunomide may prevent T-cell hyperactivation in COVID-19–related pneumonia [75].

    Teriflunomide was also suspended during hospitalization for COVID-19 in a 58-year-old woman with RRMS. Despite hospitalization, this patient developed only slight dyspnea and had adequate immunoglobulin G (IgG) antibody production by the third week of infection for both antinucleocapsid (≥1.322) and anti–spike protein S1 receptor binding domain (S1RBD; ≥1.229) [76].

    One fatality has been reported of a 55-year-old woman with advanced secondary progressive MS who interrupted teriflunomide treatment at the time of COVID-19 diagnosis. The patient had an EDSS score of 7.5 and a comorbid diagnosis of myotonic dystrophy. The patient's condition deteriorated despite being placed on supplemental oxygen, and she died 4 days after hospitalization [77].

    The consistency among these case reports suggests teriflunomide might not negatively affect the disease course of COVID-19.

    Effects on brain volume

    BVL is associated with long-term accumulation of cognitive and physical disability associated with MS, which affects patients' daily function and quality of life [25]. Additionally, disability progression occurs independent of acute focal inflammation [5] and may result from chronic smoldering disease [78]. It is therefore important that MS treatments limit BVL in addition to acute manifestations such as annualized relapses and lesion volumes.

    In vitro studies have evaluated the neuroprotective effects of teriflunomide in the CNS. In a study by Woodworth et al. [79], teriflunomide directly affected the functions of activated microglia and astrocytes. Compared with microglia treated with dimethyl sulfoxide (DMSO) control, cells from mice treated with teriflunomide exhibited suppressed proinflammatory cytokine production, reduced gene expression of proinflammatory cytokines and increased gene expression and production of interleukin 10. Additionally, stimulated astrocytes were protected against H2O2-induced cytotoxicity [79].

    Analyses of MRI data from TEMSO and TOPIC suggested teriflunomide significantly affects BVL and focal inflammation compared with placebo (Table 7). In the TEMSO core and extension study, patients with the least BVL were significantly less likely to experience disability worsening over 7 years, and BVL was predictive of long-term disability worsening at year 7 [80]. Moreover, BVL from baseline to year 2 was found to explain the greatest proportion of treatment effect of teriflunomide 14 mg on 12-week confirmed disability worsening (51.3%) compared with relapses and new or enlarging T2-weighted lesions (38.5 and 30.8%, respectively). Teriflunomide 14 mg was associated with statistically significantly smaller changes in brain volume compared with placebo in patients regardless of whether or not they had 12- or 24-week confirmed disability worsening (Figure 3) [25]. Teriflunomide was also associated with improved cognitive performance from baseline to year 2 versus placebo (p = 0.0146), and a post hoc analysis suggested the treatment effects on BVL (rather than effects on lesions or ARR) explained 44.2% of the treatment effect of teriflunomide on cognition [81].

    Table 7. Changes in brain volume following teriflunomide treatment.
    StudyPatients included, nTime frameMedicationsBrain volume measurementsOther assessmentsRef.
    BaselineChange from baseline
    Radue et al. (2017)
    TEMSO
     NCT00134563
    709 (with baseline and year 2 MRI)2 yearsTER 7 mg
     TER 14 mg
     PBO
    Normalized brain volume, cm3, mean (SD):
     TER 7 mg: 1507.8 ± 83.1
     TER 14 mg: 1502.7 ± 75.8
     PBO: 1508.9 ± 77.9
    Median percent change from baseline in brain volume:
    Year 1
     TER 7 mg: -0.4% (p = 0.0011 vs PBO)
     TER 14 mg: -0.39% (p = 0.0001 vs PBO)
     PBO: -0.61%
    Year 2
     TER 7 mg: -0.94% (p = 0.0019 vs PBO)
     TER 14 mg: -0.9% (p = 0.0001 vs PBO)
     PBO: -1.29%
    Percent change from baseline in brain volume with/without CDW, TER 14 mg vs PBO:
    With 12-week CDW
     Year 1: -0.25 vs -0.81% (p = 0.004)
     Year 2: -0.9 vs -1.61% (p = 0.004)
    With 24-week CDW
     Year 1: -0.21 vs -0.96% (p = 0.001)
     Year 2: -0.87 vs -1.78% (p = 0.006)
    Without 12-week CDW
     Year 1: -0.4 vs -0.52% (p = 0.013)
     Year 2: -0.87 vs -1.12% (p = 0.013)
    Without 24-week CDW
     Year 1: -0.41 vs -0.55% (p = 0.008)
     Year 2: -0.9 vs -1.17% (p = 0.006)
    [25]
    Sprenger et al. (2018)
    TEMSO
    NCT00134563
    522 (TER 14 mg [263] and PBO [259])2 yearsTER 14
     PBO
    NRAdjusted percent brain volume change from baseline to year 1 in patients with complete MRI data, mean (95% CI):
    Year 1
     TER: -0.41% (-0.57 to -0.26; p < 0.01 vs PBO)
     PBO: -0.69% (-0.85 to -0.53)
    Year 2
     TER: -1.01% (-1.21 to -0.81; p = 0.021 vs PBO)
     PBO: -1.30% (-1.51 to -1.09)
     [83]
    Sprenger et al. (2019)
    TOPIC
     NCT00622700
    413 (TER 14 mg [216] and PBO [197])2 yearsTER 14 mg
     PBO
    Neocortical volume, cm3, mean ± SD:
     TER: 649.4 ± 52.0
     PBO: 650.2 ± 47.8
    Adjusted annualized percent change from baseline in cortical gray matter, LS mean (95% CI):
    Year 1
     TER: -0.44% (-0.83 to -0.05; p = 0.113 vs PBO)
     PBO: -0.92% (-1.33 to -0.51)
    Year 2
     TER: -1.10% (-1.63 to -0.58; p = 0.060 vs PBO)
     PBO: -1.91% (-2.52 to -1.29)
     [90]
    Wuerfel et al. (2019)
    TEMSO
     NCT00134563
    709 (with baseline MRI and 108-week follow-up)2 yearsTER 14 mg
     PBO
     LS mean difference (95% CI) in BVL from baseline to Year 2: 0.46% (0.16–0.76; p = 0.0008) [81]
    Sprenger et al. (2020)
    TEMSO
     NCT00803049
    709 (Group 1/least BVL [221], Group 2/intermediate BVL [357] and Group 3/most BVL [131])Up to 7 yearsTER 7 mg TER 14 mg
     PBO
    NRLS mean change in BVL from baseline to year 2 was 0.46% (95% CI, 0.16 to 0.76%) lower with TER vs PBO (p < 0.001)Patients with minimal or intermediate BVL (≤0.52 or >0.52 to 2.18%) from baseline to year 2 were significantly less likely than those with most BVL (>2.18%) to have CDW at year 7:
     12-week CDW: HRs 0.58–0.68 (p < 0.01 for minimal or intermediate BVL vs high BVL)
     24-week CDW: HRs 0.57–0.71 (p < 0.05 for minimal or intermediate BVL vs high BVL)
     BVL at year 2 accounted for 51.3% of treatment effect on CDW
    [80]
    Zivadinov et al. (2020)
    TOPIC
     NCT00622700
    485 (TER 7 mg [156], TER 14 mg [170] and PBO [159])Up to 4 yearsTER 7 mg
     TER 14 mg
     PBO
    Normalized gray matter volume, mL, mean ± SD:
     TER 7 mg: 651.0 ± 56.6
     TER 14 mg: 648.0 ± 51.4
     PBO: 648.0 ± 47.6
    Median percent change from baseline in cortical gray matter volume:
    Month 6
     TER 7 mg: -0.16% (p = 0.0937 vs PBO)
     TER 14 mg: +0.07% (p = 0.0185 vs PBO)
     PBO: -0.38%
    Month 12
     TER 7 mg: -0.17% (p = 0.0543 vs PBO)
     TER 14 mg: -0.32% (p = 0.0359 vs PBO)
     PBO: -0.83%
    Month 18
     TER 7 mg: -0.41% (p = 0.0035 vs PBO)
     TER 14 mg: -0.45% (p = 0.0027 vs PBO)
     PBO: -1.35%
    Month 24
     TER 7 mg: -0.76% (p = 0.0087 vs PBO)
     TER 14 mg: -0.84% (p = 0.0416 vs PBO)
     PBO: -1.41%
     [26]
    Sprenger et al. (2019)
    TOPIC
     NCT00622700
    413 (TER 14 mg [216] and PBO [197])2 yearsTER 14 mg
     PBO
    Normalized brain volume, cm3, mean ± SD:
     TER: 1594.4 ± 89.5
     PBO: 1592.2 ± 85.7
    Adjusted percent brain volume change from baseline, mean (95% CI):
    Year 1
     TER: -0.24% (-0.34 to -0.13; p = 0.141 vs PBO)
     PBO: -0.30% (-0.40 to -0.19)
    Year 2
     TER: -0.62% (-0.70 to -0.54; p < 0.0001 vs PBO)
     PBO: -0.79% (-0.88 to -0.70)
     [91]
    Hauser et al. (2020)
     ASCLEPIOS I and II NCT02792218
     NCT02792231
    ASCLEPIOS I: N = 927
     ASCLEPIOS II: N = 955
    Up to 30 monthsTER 14 mg QD
     OFA 20 mg every 4 weeks
    Normalized brain volume, cm3, mean ± SD:
    ASCLEPIOS I
     TER: 1442 ± 79
     OFA: 1439 ± 81
    ASCLEPIOS II
     TER: 1446 ± 77
     OFA: 1441 ± 77
    Annual rate of brain volume change (95% CI):
    ASCLEPIOS I
     TER: -0.35% (-0.41 to -0.29; p = 0.12 vs OFA)
     OFA: -0.28% (-0.34 to -0.22)
    ASCLEPIOS II
     TER: -0.35% (-0.42 to -0.29; p = 0.13 vs OFA)
     OFA: -0.29% (-0.35 to -0.23)
     [46]
    Zivadinov et al. (2019)1009–24 monthsTER 14 mg QD
     DMF 240 mg BID
    NRMedian (Q1–Q3) annualized percent change in whole-brain volume:
     TER: -0.1% (-0.5 to 0.4; p = 0.0212 vs DMF)
     DMF: -0.5% (-0.9 to -0.2)
    Median (Q1–Q3) annualized percent lateral ventricular volume change:
     TER: 1.2% (-1.7 to 4.5; p = 0.2874)
     DMF: 3.0% (-0.4 to 6.0)
    Proportion of patients who developed brain atrophy (percent whole-brain volume change <-0.4%):
     TER: 25.9% (p = 0.0135 vs DMF)
    DMF: 58.6%
    [82]

    BID: Twice daily; BVL: Brain volume loss; CDW: Confirmed disability worsening; CI: Confidence interval; DMF: Dimethyl fumarate; HR: Hazard ratio; LS: Least squares; MRI: Magnetic resonance imaging; NR: Not reported; OFA: Ofatumumab; PBO: Placebo; QD: Once daily; SD: Standard deviation; TER: Teriflunomide.

    Figure 3. Brain volume loss according to on-study disability worsening in the Phase III TEMSO trial.

    (A) BVL in placebo-treated patients with and without 12- or 24-week CDW. (B) BVL in placebo- and teriflunomide 14 mg–treated patients with 12- or 24-week CDW. (C) BVL in placebo- and teriflunomide 14 mg–treated patients without 12- or 24-week CDW.

    *Relative change versus patients without CDW.

    **Relative change versus placebo.

    BVL: Brain volume loss; CDW: Confirmed disability worsening.

    Reproduced from Radue et al. [25].

    In TOPIC, teriflunomide 14 mg was associated with significantly smaller changes in cortical gray matter volume versus placebo at months 6–24 [26]. Also, greater magnitude of cortical gray matter BVL was associated with a higher risk of conversion to clinically definite MS [26]. ASCLEPIOS I and II reported similar annual rates of BVL between teriflunomide and ofatumumab, based on assessments of percent brain volume change from baseline performed at 12 and 24 months [46]. In comparison, the real-world Teri-RADAR study described significantly reduced annualized percent whole BVL at 24 months compared with DMF (median change, -0.1 vs -0.5; p = 0.0212) [82]. The consistency of these data suggest teriflunomide may reduce BVL in patients with relapsing forms of MS, which is corroborated in the consistent effects seen in reduction of disability worsening and improvement of quality-of-life measures. Future efforts should explore whether these observations are potentially related to teriflunomide's effect on diffuse and/or chronic smoldering inflammation [83].

    Regulatory affairs

    Teriflunomide is approved in more than 80 countries as of January 2021. US approval for the treatment of relapsing forms of MS includes clinically isolated syndrome, RRMS and active secondary progressive disease, in adults. Both 7- and 14-mg doses are approved in the US, whereas approval in other countries is only for the 14-mg dose. In the EU, teriflunomide is indicated for the treatment of adults with RRMS.

    Conclusion

    Clinical trials of teriflunomide treatment lasting up to 9 years in adults with relapsing forms of MS or with a first clinical episode suggestive of MS consistently demonstrated lasting improvements in disease measures such as ARR, disability worsening and MRI outcomes. Data from the clinical development program have now been supplemented by real-world evidence and comparisons with other DMTs in patients with relapsing forms of MS. Treatment safety and tolerability in these studies has been consistent with that seen in the clinical development program, further supporting the long-term safety and tolerability of teriflunomide. The pediatric TERIKIDS recently reported efficacy and safety results similar to those seen in adult studies, in which time to relapse and MRI activity were reduced with teriflunomide versus placebo with no unexpected safety signals.

    Consistently, teriflunomide has demonstrated a significant impact on disability progression and brain atrophy across clinical trials and real-world studies starting early in the disease course. This suggests teriflunomide targets the neurodegenerative process in addition to the well-characterized effect on focal inflammation.

    Patient-reported outcomes from Teri-PRO and other observational studies suggest high overall patient satisfaction with teriflunomide, both in patients new to DMTs and in those who switch from another DMT. Patients previously treated with other DMTs report particularly high treatment satisfaction related to convenience and side effects.

    Preliminary evidence suggests teriflunomide supports an immune antiviral response, in contrast to other DMTs that may increase susceptibility for viral infections. Initial case reports of patients infected with COVID-19 also suggest teriflunomide does not negatively affect the disease course.

    Taken together, these results from recent clinical trial and real-world analyses add to the established safety and efficacy profiles for teriflunomide, further supporting its use in treating relapsing forms of MS.

    Future perspective

    DMTs have been shown to reduce the risk of focal inflammation, but do not necessarily affect long-term disease progression or patient disability. For many patients, disease continues to progress despite the use of DMTs to control relapses and lesions. Therapeutic options are needed that can address the chronic smoldering inflammation that remains ongoing during remissions, which plays a role in progression of the disease. Given the potential for such therapies to improve quality of life and long-term outcomes for patients with relapsing forms of MS, research may increasingly focus on the pathological drivers of chronic smoldering inflammation in MS and the ability of current or new DMTs to target this underlying disease activity.

    Author contributions

    Aaron E Miller fulfilled the ICMJE criteria for authorship, including the approval of the final version for submission.

    Acknowledgments

    Critical review of the manuscript was provided by Svend Geertsen, PhD, Pascal Rufi, MD and Michael Yeakey, PharmD, of Sanofi.

    Financial & competing interests disclosure

    AE Miller reports having received consulting fees from AbbVie, Accordant Health Services (CVS Caremark), Adamas, Biogen, Bristol Myers Squibb (Celgene), EMD Serono, Genentech/Roche, Greenwich Bioscience, Janssen, Mapi Pharma, Mylan, Novartis, Sanofi/Genzyme and Verana Health; speaking fees (disease awareness programs only) from Alexion, Biogen and Genentech; and contracted research from Genentech, MedDay, Novartis, Roche and Sanofi. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Editorial support (assistance in drafting and editing of the manuscript text, figures and tables, as directed by the author, data checking and incorporation of comments from reviewers, and assisting with the submission process) was provided by P Xenopoulos and A Kelly of Elevate Scientific Solutions, funded by Sanofi.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Dobson R, Giovannoni G. Multiple sclerosis – a review. Eur. J. Neurol. 26(1), 27–40 (2019).
    • 2. Multiple Sclerosis International Federation. Atlas of MS (3rd Edition). (2020). https://www.atlasofms.org/map/global/epidemiology/number-of-people-with-ms
    • 3. Thompson AJ, Baranzini SE, Geurts J, Hemmer B, Ciccarelli O. Multiple sclerosis. Lancet 391(10130), 1622–1636 (2018).
    • 4. Miller AE. Teriflunomide in multiple sclerosis: an update. Neurodegener. Dis. Manag. 7(1), 9–29 (2017).
    • 5. University of California San Francisco MS Epic Team, Cree BAC, Hollenbach JA et al. Silent progression in disease activity-free relapsing multiple sclerosis. Ann. Neurol. 85(5), 653–666 (2019).
    • 6. Betaseron (interferon beta-1b) for injection [prescribing information]. Bayer HealthCare Pharmaceuticals Inc, NJ, USA (2019).
    • 7. Extavia (interferon beta-1b) for injection [prescribing information]. Novartis Pharmaceuticals Corp, NJ, USA (2019).
    • 8. Avonex (interferon beta-1a) injection [prescribing information]. Biogen Inc, MA, USA (2020).
    • 9. Rebif (interferon beta-1a) [prescribing information]. EMD Serono Inc, MA, USA (2020).
    • 10. Plegridy (peginterferon beta-1a) injection [prescribing information]. Biogen Inc, MA, USA (2020).
    • 11. Copaxone (glatiramer acetate injection) [prescribing information]. Teva Neuroscience Inc, NJ, USA (2020).
    • 12. Gilenya (fingolimod) capsules [prescribing information]. Novartis Pharmaceuticals Corp, NJ, USA (2019).
    • 13. Zeposia (ozanimod) capsules [prescribing information]. Celgene Corp, NJ, USA (2020).
    • 14. Mayzent (siponimod) tablets [prescribing information]. Novartis Pharmaceuticals Corp, NJ, USA (2019).
    • 15. Tecfidera (dimethyl fumarate) delayed-release capsules [prescribing information]. Biogen Inc, MA, USA (2019).
    • 16. Vumerity (diroximel fumarate) delayed-release capsules [prescribing information]. Alkermes Inc, MA, USA (2019).
    • 17. Bafiertam (monomethyl fumarate) delayed-release capsules [prescribing information]. Banner Life Sciences, LLC, NC, USA (2020).
    • 18. Mavenclad (cladribine) tablets [prescribing information]. EMD Serono Inc, MA, USA (2019).
    • 19. Tysabri (natalizumab) injection [prescribing information]. Biogen Inc, MA, USA (2020).
    • 20. Lemtrada (alemtuzumab) [prescribing information]. Genzyme Corp, MA, USA (2020).
    • 21. Ocrevus (ocrelizumab) injection [prescribing information]. Genentech Inc, CA, USA (2020).
    • 22. Kesimpta (ofatumumab) [prescribing information]. Novartis Pharmaceuticals Corp, NJ, USA (2020).
    • 23. Aubagio (teriflunomide) [prescribing information]. Genzyme Corp, MA, USA (2019).
    • 24. Aubagio (teriflunomide) summary of product characteristics. Sanofi-Aventis Groupe, Paris, France (2018).
    • 25. Radue EW, Sprenger T, Gaetano L et al. Teriflunomide slows BVL in relapsing MS: a reanalysis of the TEMSO MRI data set using SIENA. Neurol. Neuroimmunol. Neuroinflamm. 4(5), e390 (2017).
    • 26. Zivadinov R, Dwyer MG, Carl E et al. Slowing of brain atrophy with teriflunomide and delayed conversion to clinically definite MS. Ther. Adv. Neurol. Disord. 13, 1756286420970754 (2020). •• During 2-year treatment with teriflunomide in the Phase III TOPIC study, rates of cortical gray matter and whole-brain atrophy were reduced compared with placebo. Every 1% reduction in brain volume was associated with a corresponding 14.5% (gray matter) and 47.3% (whole brain) increased risk of conversion from clinically isolated syndrome to clinically definite multiple sclerosis (MS).
    • 27. O'Connor P, Wolinsky JS, Confavreux C et al. Randomized trial of oral teriflunomide for relapsing multiple sclerosis. N. Engl. J. Med. 365(14), 1293–1303 (2011).
    • 28. Claussen MC, Korn T. Immune mechanisms of new therapeutic strategies in MS: teriflunomide. Clin. Immunol. 142(1), 49–56 (2012).
    • 29. Wiese MD, Rowland A, Polasek TM, Sorich MJ, O'Doherty C. Pharmacokinetic evaluation of teriflunomide for the treatment of multiple sclerosis. Expert Opin. Drug Metab. Toxicol. 9(8), 1025–1035 (2013).
    • 30. Bar-Or A, Pachner A, Menguy-Vacheron F, Kaplan J, Wiendl H. Teriflunomide and its mechanism of action in multiple sclerosis. Drugs 74(6), 659–674 (2014).
    • 31. Wiendl H, Gross CC, Lindner M et al. Characterizing the impact of teriflunomide on adaptive immune cell subsets, repertoire and function in patients with relapsing remitting MS: TERI-DYNAMIC. Presented at: 31st Congress of the European Committee for Treatment and Research in Multiple Sclerosis. Barcelona, Spain, 7–10 October 2015.
    • 32. Comi G, Miller AE, Benamor M, Truffinet P, Poole EM, Freedman MS. Characterizing lymphocyte counts and infection rates with long-term teriflunomide treatment: pooled analysis of clinical trials. Mult. Scler. 26(9), 1083–1092 (2020).
    • 33. Bar-Or A, Wiendl H, Miller B et al. Randomized study of teriflunomide effects on immune responses to neoantigen and recall antigens. Neurol. Neuroimmunol. Neuroinflamm. 2(2), e70 (2015).
    • 34. Bar-Or A, Freedman MS, Kremenchutzky M et al. Teriflunomide effect on immune response to influenza vaccine in patients with multiple sclerosis. Neurology 81(6), 552–558 (2013).
    • 35. Prinz C, Millward JM, Rothe M et al. Investigating the biodistribution of the anti-inflammatory drug teriflunomide in vivo using 19F MRI and mass spectrometry. Presented at: 35th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Stockholm, Sweden, 11–13 September 2019.
    • 36. Kaplan J, Cavalier S, Turpault S. Biodistribution of teriflunomide in naïve rats vs rats with experimental autoimmune encephalomyelitis. Presented at: 31st Congress of the European Committee for Treatment and Research in Multiple Sclerosis. Barcelona, Spain, 7–10 October 2015.
    • 37. Kremenchutzky M, Freedman M, Bar-Or A et al. Final outcomes of the teriflunomide Phase 2 extension study: 13 years of efficacy and safety results (P3.027). Neurology 86(Suppl. 16), P3.027 (2016).
    • 38. O'Connor PW, Li D, Freedman MS et al. A Phase II study of the safety and efficacy of teriflunomide in multiple sclerosis with relapses. Neurology 66(6), 894–900 (2006).
    • 39. Confavreux C, O'Connor P, Comi G et al. Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, Phase 3 trial. Lancet Neurol. 13(3), 247–256 (2014).
    • 40. Miller AE, Wolinsky JS, Kappos L et al. Oral teriflunomide for patients with a first clinical episode suggestive of multiple sclerosis (TOPIC): a randomised, double-blind, placebo-controlled, Phase 3 trial. Lancet Neurol. 13(10), 977–986 (2014).
    • 41. Vermersch P, Czlonkowska A, Grimaldi LM et al. Teriflunomide versus subcutaneous interferon beta-1a in patients with relapsing multiple sclerosis: a randomised, controlled Phase 3 trial. Mult. Scler. 20(6), 705–716 (2014).
    • 42. Chitnis T, Banwell B, Kappos L et al. TERIKIDS study: teriflunomide efficacy and safety in paediatric patients with relapsing forms of MS. Presented at: 6th Congress of the European Academy of Neurology (EAN). Virtual, 23–26 May 2020).
    • 43. Chitnis T, Banwell B, Arnold DL et al. Teriflunomide efficacy and safety in pediatric patients with relapsing forms of MS: interim analysis of open-label terikids trial extension. Abstract FC02.04. Mult. Scler. 26(Suppl. 3), 5–6 (2020).
    • 44. Vermersch P, Gold R, Meca-Lallana JE et al. Treatment satisfaction with teriflunomide in patients switching from a prior disease-modifying therapy: results from the Phase 3 TENERE extension and Phase 4 Teri-PRO clinical trials. Presented at: 70th Annual Meeting of the American Academy of Neurology. CA, USA, 21–27 April 2018.
    • 45. Kappos L, Burcklen M, Freedman M et al. Efficacy and safety of ponesimod compared to teriflunomide in patients with relapsing multiple sclerosis: results of the randomized, active-controlled, double-blind, parallel-group Phase 3 OPTIMUM study. Presented at: 35th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Stockholm, Sweden, 11–13 September 2019.
    • 46. Hauser SL, Bar-Or A, Cohen JA et al. Ofatumumab versus teriflunomide in multiple sclerosis. N. Engl. J. Med. 383(6), 546–557 (2020). • In the Phase III ASCLEPIOS I and II trials, ofatumumab treatment was associated with reduced annualized relapse rates, MRI lesions and rates of disability worsening versus teriflunomide, but similar rates of clinical disability improvement and BVL were observed with both treatments.
    • 47. Freedman MS, Montalban X, Miller AE et al. Comparing outcomes from clinical studies of oral disease-modifying therapies (dimethyl fumarate, fingolimod, and teriflunomide) in relapsing MS: assessing absolute differences using a number needed to treat analysis. Mult. Scler. Relat. Disord. 10, 204–212 (2016).
    • 48. Coyle PK, Khatri B, Edwards KR et al. Patient-reported outcomes in patients with relapsing forms of MS switching to teriflunomide from other disease-modifying therapies: results from the global Phase 4 Teri-PRO study in routine clinical practice. Mult. Scler. Relat. Disord. 26, 211–218 (2018). • Teri-PRO was a global, open-label, Phase IV study conducted in routine clinical practice. Patients with relapsing forms of MS reported improved treatment satisfaction and improved or stable disability worsening, cognition and quality of life after switching from another DMT to teriflunomide.
    • 49. Coyle PK, Khatri B, Edwards KR et al. Patient-reported outcomes in relapsing forms of MS: real-world, global treatment experience with teriflunomide from the Teri-PRO study. Mult. Scler. Relat. Disord. 17, 107–115 (2017). • In Teri-PRO, patients with relapsing forms of MS received teriflunomide as an initial DMT or after switching from another DMT. The overall population reported high global treatment satisfaction after 48 weeks of teriflunomide treatment, with the highest satisfaction scores seen for side effects and convenience. Self-reported quality of life improved and cognition was stable over 48 weeks.
    • 50. Gobbi C, Achtnichts L, Derfuss T et al. Real world data from the TACO (teriflunomide in relapsing remitting multiple sclerosis patients assessing clinical benefit and patient reported outcomes) study. Presented at: 35th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Stockholm, Sweden, 11–13 September 2019.
    • 51. Kallmann BA, Tiel-Wilck K, Kullmann JS, Engelmann U, Chan A. Real-life outcomes of teriflunomide treatment in patients with relapsing multiple sclerosis: TAURUS-MS observational study. Ther Adv Neurol Disord 12, 1756286419835077 (2019). • In the observational TAURUS-MS study, 2-year treatment with teriflunomide led to reductions in annualized relapse rate, stable Expanded Disability Status Scale and patient-reported fatigue, and increased overall treatment satisfaction.
    • 52. de Seze J, Devy R, Planque E et al. Fatigue in teriflunomide-treated patients with relapsing remitting multiple sclerosis in the real-world Teri-FAST study. Mult. Scler. Relat. Disord. 47, 102659 (2020).
    • 53. Menascu S, Petrou P, Karussis D et al. Real-world safety, health, and patient-reported outcomes after teriflunomide 14 mg treatment initiation in patients with relapsing remitting multiple sclerosis: teri-REAL. Presented at: 35th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Stockholm, Sweden, 11–13 September 2019.
    • 54. Hestvik AL, Frederiksen J, Nielsen HH et al. Teri-LIFE: an observational study of quality of life in patients with relapsing remitting multiple sclerosis treated with teriflunomide in the Nordic region. Presented at: 35th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Stockholm, Sweden, 11–13 September 2019.
    • 55. Comi G, Freedman MS, Kappos L et al. Pooled safety and tolerability data from four placebo-controlled teriflunomide studies and extensions. Mult. Scler. Relat. Disord. 5, 97–104 (2016).
    • 56. Vukusic S, Coyle PK, Jurgensen S et al. Pregnancy outcomes in patients with multiple sclerosis treated with teriflunomide: clinical study data and 5 years of post-marketing experience. Mult. Scler. 26(7), 829–836 (2020).
    • 57. Vukusic S, Hellwig K, Truffinet P et al. Pregnancy outcomes in female partners of male patients treated with teriflunomide or leflunomide (the parent compound of teriflunomide). Presented at: 35th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Stockholm, Sweden, 11–13 September 2019.
    • 58. Andersen JB, Moberg JY, Spelman T, Magyari M. Pregnancy outcomes in men and women treated with teriflunomide. A population-based nationwide Danish register study. Front. Immunol. 9, 2706 (2018).
    • 59. McKay KA, Kwan V, Duggan T, Tremlett H. Risk factors associated with the onset of relapsing-remitting and primary progressive multiple sclerosis: a systematic review. Biomed. Res. Int. 2015, 817238 (2015).
    • 60. Krone B, Oeffner F, Grange JM. Is the risk of multiple sclerosis related to the ‘biography’ of the immune system? J. Neurol. 256(7), 1052–1060 (2009).
    • 61. Winkelmann A, Loebermann M, Reisinger EC, Hartung HP, Zettl UK. Disease-modifying therapies and infectious risks in multiple sclerosis. Nat. Rev. Neurol. 12(4), 217–233 (2016).
    • 62. Virtanen JO, Jacobson S. Viruses and multiple sclerosis. CNS Neurol. Disord. Drug Targets 11(5), 528–544 (2012).
    • 63. Saribas AS, Ozdemir A, Lam C, Safak M. JC virus-induced progressive multifocal leukoencephalopathy. Future Virol. 5(3), 313–323 (2010).
    • 64. Yong KP, Kim HJ. Disease modifying therapies and infection risks in multiple sclerosis-a decision-making conundrum. Ann. Transl. Med. 8(11), 722 (2020).
    • 65. Edwards KR, Turner TJ, Able RA, Mandel M. Potential antiviral properties of teriflunomide: a consideration for optimizing MS treatment sequencing. Presented at: Americas Committee for Treatment and Research in Multiple Sclerosis (ACTRIMS) Forum. FA, USA, 23–25 February 2017.
    • 66. Atwood WJ, Turner T, Chavin J, Guikema B, O'Hara B. Effect of teriflunomide on JCPyV infection in primary astrocytes and choroid plexus epithelial cells. Presented at: 35th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Stockholm, Sweden, 11–13 September 2019.
    • 67. Berger JR, Aksamit AJ, Clifford DB et al. PML diagnostic criteria: consensus statement from the AAN Neuroinfectious Disease Section. Neurology 80(15), 1430–1438 (2013).
    • 68. Sanofi. Pharmacovigilance data on file (31 October 2020).
    • 69. Akahata Y, Ngouth N, Ohayon J et al. Teriflunomide reduces spontaneous lymphoproliferation of peripheral blood mononuclear cells from patients with human t-lymphotropic virus (HTLV)-1-associated myelopathy/tropical spastic paraparesis. Presented at: 34th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Berlin, Germany, 10–12 October 2018.
    • 70. Gold J, Holden D, Parratt J, Yiannikas C, Ahmad R, Giovannoni G. Effect of teriflunomide on Epstein-Barr virus shedding in relapsing-remitting multiple sclerosis patients: outcomes from a real-world cohort study. Presented at: 8th Joint European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS) – Americas Committee for Treatment and Research in Multiple Sclerosis (ACTRIMS) Meeting. DC, USA, September 11–13 2020.
    • 71. Zivadinov R, Ramanathan M, Hagemeier J et al. Teriflunomide's effect on humoral response to Epstein-Barr virus and development of cortical gray matter pathology in multiple sclerosis. Mult. Scler. Relat. Disord. 36, 101388 (2019).
    • 72. Maghzi AH, Houtchens MK, Preziosa P et al. COVID-19 in teriflunomide-treated patients with multiple sclerosis. J. Neurol. 267(10), 2790–2796 (2020).
    • 73. Mantero V, Baroncini D, Balgera R et al. Mild COVID-19 infection in a group of teriflunomide-treated patients with multiple sclerosis. J. Neurol. 268 (6), 2029–2030 (2020).
    • 74. Möhn N, Saker F, Bonda V et al. Mild COVID-19 symptoms despite treatment with teriflunomide and high-dose methylprednisolone due to multiple sclerosis relapse. J. Neurol. 267(10), 2803–2805 (2020).
    • 75. Ciardi MR, Zingaropoli MA, Pasculli P et al. The peripheral blood immune cell profile in a teriflunomide-treated multiple sclerosis patient with COVID-19 pneumonia. J. Neuroimmunol. 346, 577323 (2020).
    • 76. Bollo L, Guerra T, Bavaro DF et al. Seroconversion and indolent course of COVID-19 in patients with multiple sclerosis treated with fingolimod and teriflunomide. J. Neurol. Sci. 416, 117011 (2020).
    • 77. Bowen JD, Brink J, Brown TR et al. COVID-19 in MS: initial observations from the Pacific Northwest. Neurol. Neuroimmunol. Neuroinflamm. 7(5), e783 (2020).
    • 78. Absinta M, Sati P, Masuzzo F et al. Association of chronic active multiple sclerosis lesions with disability in vivo. JAMA Neurol. 76(12), 1474–1483 (2019).
    • 79. Woodworth L, Mahan A, Garron T et al. In vitro data reveals potential novel mechanism of action of teriflunomide on CNS microglia and astrocytes. Presented at: 7th Joint European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS) – Americas Committee for Treatment and Research in Multiple Sclerosis (ACTRIMS) Meeting. Paris, France, 25–28 October 2017.
    • 80. Sprenger T, Kappos L, Radue EW et al. Association of brain volume loss and long-term disability outcomes in patients with multiple sclerosis treated with teriflunomide. Mult. Scler. 26(10), 1207–1216 (2020). •• Analysis of BVL from patients in TEMSO (core and extension studies) suggested significant reductions in disability worsening at year 7 for patients with the least versus the most BVL. The treatment effect on disability worsening was primarily explained by brain volume loss at year 2 (51.3%), but also by relapses during the first 2 years (38.5%) and new/enlarging T2-weighted lesions over 2 years (30.8%).
    • 81. Wuerfel J, Macdonnell R, Sormani MP et al. Brain volume loss and cognition in teriflunomide-treated patients in TEMSO. Presented at: 71st Annual Meeting of the American Academy of Neurology (AAN). PA, USA, 4–10 May 2019.
    • 82. Zivadinov R, Kresa-Reahl K, Weinstock-Guttman B et al. Comparative effectiveness of teriflunomide and dimethyl fumarate in patients with relapsing forms of MS in the retrospective real-world Teri-RADAR study. J. Comp. Eff. Res. 8(5), 305–316 (2019).
    • 83. Sprenger T, Comi G, Lebrun-Frenay C et al. Investigating the effect of teriflunomide on diffuse brain tissue damage in the Phase 3 TEMSO Study. 34th Congress of the European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS). Berlin, Germany, 10–12 October 2018.
    • 84. Glatopa (glatiramer acetate injection) [prescribing information]. Novartis AG, NJ, USA, 2018.
    • 85. Novantrone (mitoxantrone) [prescribing information]. Hospira Inc, IL, USA, 2021.
    • 86. Ponvory (ponesimod) [prescribing information]. Janssen Pharmaceuticals Inc, NJ, USA, 2021.
    • 87. O'Connor P, Comi G, Freedman MS et al. Long-term safety and efficacy of teriflunomide: nine-year follow-up of the randomized TEMSO study. Neurology 86(10), 920–930 (2016).
    • 88. Miller AE, Olsson TP, Wolinsky JS et al. Long-term safety and efficacy of teriflunomide in patients with relapsing multiple sclerosis: results from the TOWER extension study. Mult. Scler. Relat. Disord. 46, 102438 (2020).
    • 89. Miller AE, Vermersch P, Kappos L et al. Long-term outcomes with teriflunomide in patients with clinically isolated syndrome: results of the TOPIC extension study. Mult. Scler. Relat. Disord. 33, 131–138 (2019).
    • 90. Sprenger T, Lebrun-Frenay C, Vermersch P et al. Investigating the effect of teriflunomide on diffuse cortical grey matter volume loss in the Phase 3 TOPIC study. Presented at: 2019 Annual Meeting of the European Academy of Neurology (EAN). Oslo, Norway, 29 June–2 July 2019.
    • 91. Sprenger T, Lebrun-Frenay C, Vermersch P et al. Investigating the effect of teriflunomide on diffuse brain atrophy independent of focal disease activity in the Phase 3 TOPIC study. Presented at: 71st Annual Meeting of the American Academy of Neurology (AAN). PA, USA, 4–10 May 2019.